Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Discov ; 9(1): 32, 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36964131

ABSTRACT

Calpains are a class of non-lysosomal cysteine proteases that exert their regulatory functions via limited proteolysis of their substrates. Similar to the lysosomal and proteasomal systems, calpain dysregulation is implicated in the pathogenesis of neurodegenerative disease and cancer. Despite intensive efforts placed on the identification of mechanisms that regulate calpains, however, calpain protein modifications that regulate calpain activity are incompletely understood. Here we show that calpains are regulated by KCTD7, a cytosolic protein of previously uncharacterized function whose pathogenic mutations result in epilepsy, progressive ataxia, and severe neurocognitive deterioration. We show that KCTD7 works in complex with Cullin-3 and Rbx1 to execute atypical, non-degradative ubiquitination of calpains at specific sites (K398 of calpain 1, and K280 and K674 of calpain 2). Experiments based on single-lysine mutants of ubiquitin determined that KCTD7 mediates ubiquitination of calpain 1 via K6-, K27-, K29-, and K63-linked chains, whereas it uses K6-mediated ubiquitination to modify calpain 2. Loss of KCTD7-mediated ubiquitination of calpains led to calpain hyperactivation, aberrant cleavage of downstream targets, and caspase-3 activation. CRISPR/Cas9-mediated knockout of Kctd7 in mice phenotypically recapitulated human KCTD7 deficiency and resulted in calpain hyperactivation, behavioral impairments, and neurodegeneration. These phenotypes were largely prevented by pharmacological inhibition of calpains, thus demonstrating a major role of calpain dysregulation in KCTD7-associated disease. Finally, we determined that Cullin-3-KCTD7 mediates ubiquitination of all ubiquitous calpains. These results unveil a novel mechanism and potential target to restrain calpain activity in human disease and shed light on the molecular pathogenesis of KCTD7-associated disease.

2.
Small GTPases ; 13(1): 14-47, 2022 01.
Article in English | MEDLINE | ID: mdl-33955328

ABSTRACT

Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.


Subject(s)
Autism Spectrum Disorder , rho GTP-Binding Proteins , Humans , rho GTP-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Autism Spectrum Disorder/metabolism , Synapses/metabolism , Signal Transduction
3.
J Neurosci ; 41(6): 1191-1206, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33328293

ABSTRACT

The dentate gyrus (DG) controls information flow into the hippocampus and is critical for learning, memory, pattern separation, and spatial coding, while DG dysfunction is associated with neuropsychiatric disorders. Despite its importance, the molecular mechanisms regulating DG neural circuit assembly and function remain unclear. Here, we identify the Rac-GEF Tiam1 as an important regulator of DG development and associated memory processes. In the hippocampus, Tiam1 is predominantly expressed in the DG throughout life. Global deletion of Tiam1 in male mice results in DG granule cells with simplified dendritic arbors, reduced dendritic spine density, and diminished excitatory synaptic transmission. Notably, DG granule cell dendrites and synapses develop normally in Tiam1 KO mice, resembling WT mice at postnatal day 21 (P21), but fail to stabilize, leading to dendrite and synapse loss by P42. These results indicate that Tiam1 promotes DG granule cell dendrite and synapse stabilization late in development. Tiam1 loss also increases the survival, but not the production, of adult-born DG granule cells, possibly because of greater circuit integration as a result of decreased competition with mature granule cells for synaptic inputs. Strikingly, both male and female mice lacking Tiam1 exhibit enhanced contextual fear memory and context discrimination. Together, these results suggest that Tiam1 is a key regulator of DG granule cell stabilization and function within hippocampal circuits. Moreover, based on the enhanced memory phenotype of Tiam1 KO mice, Tiam1 may be a potential target for the treatment of disorders involving memory impairments.SIGNIFICANCE STATEMENT The dentate gyrus (DG) is important for learning, memory, pattern separation, and spatial navigation, and its dysfunction is associated with neuropsychiatric disorders. However, the molecular mechanisms controlling DG formation and function remain elusive. By characterizing mice lacking the Rac-GEF Tiam1, we demonstrate that Tiam1 promotes the stabilization of DG granule cell dendritic arbors, spines, and synapses, whereas it restricts the survival of adult-born DG granule cells, which compete with mature granule cells for synaptic integration. Notably, mice lacking Tiam1 also exhibit enhanced contextual fear memory and context discrimination. These findings establish Tiam1 as an essential regulator of DG granule cell development, and identify it as a possible therapeutic target for memory enhancement.


Subject(s)
Dendrites/metabolism , Dentate Gyrus/metabolism , Memory/physiology , Neurogenesis/physiology , Synapses/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1/deficiency , Animals , Dendrites/genetics , Dentate Gyrus/cytology , Female , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Synapses/genetics , T-Lymphoma Invasion and Metastasis-inducing Protein 1/genetics
4.
Nature ; 598(7879): 144-150, 2021 10.
Article in English | MEDLINE | ID: mdl-33184512

ABSTRACT

Cortical neurons exhibit extreme diversity in gene expression as well as in morphological and electrophysiological properties1,2. Most existing neural taxonomies are based on either transcriptomic3,4 or morpho-electric5,6 criteria, as it has been technically challenging to study both aspects of neuronal diversity in the same set of cells7. Here we used Patch-seq8 to combine patch-clamp recording, biocytin staining, and single-cell RNA sequencing of more than 1,300 neurons in adult mouse primary motor cortex, providing a morpho-electric annotation of almost all transcriptomically defined neural cell types. We found that, although broad families of transcriptomic types (those expressing Vip, Pvalb, Sst and so on) had distinct and essentially non-overlapping morpho-electric phenotypes, individual transcriptomic types within the same family were not well separated in the morpho-electric space. Instead, there was a continuum of variability in morphology and electrophysiology, with neighbouring transcriptomic cell types showing similar morpho-electric features, often without clear boundaries between them. Our results suggest that neuronal types in the neocortex do not always form discrete entities. Instead, neurons form a hierarchy that consists of distinct non-overlapping branches at the level of families, but can form continuous and correlated transcriptomic and morpho-electrical landscapes within families.


Subject(s)
Gene Expression Profiling , Motor Cortex/cytology , Neurons/classification , Neurons/metabolism , Transcriptome , Animals , Atlases as Topic , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Glutamates/metabolism , Lysine/analogs & derivatives , Lysine/analysis , Male , Mice , Motor Cortex/anatomy & histology , Neurons/cytology , Organ Specificity , Patch-Clamp Techniques , Phenotype , Sequence Analysis, RNA , Single-Cell Analysis , Staining and Labeling
5.
Elife ; 92020 03 05.
Article in English | MEDLINE | ID: mdl-32134385

ABSTRACT

Clones of excitatory neurons derived from a common progenitor have been proposed to serve as elementary information processing modules in the neocortex. To characterize the cell types and circuit diagram of clonally related excitatory neurons, we performed multi-cell patch clamp recordings and Patch-seq on neurons derived from Nestin-positive progenitors labeled by tamoxifen induction at embryonic day 10.5. The resulting clones are derived from two radial glia on average, span cortical layers 2-6, and are composed of a random sampling of transcriptomic cell types. We find an interaction between shared lineage and connection type: related neurons are more likely to be connected vertically across cortical layers, but not laterally within the same layer. These findings challenge the view that related neurons show uniformly increased connectivity and suggest that integration of vertical intra-clonal input with lateral inter-clonal input may represent a developmentally programmed connectivity motif supporting the emergence of functional circuits.


Subject(s)
Neocortex/cytology , Neurons/classification , Neurons/physiology , Synapses/physiology , Animals , Cells, Cultured , Mice
6.
Cells ; 9(1)2020 01 18.
Article in English | MEDLINE | ID: mdl-31963704

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. TBIs, which range in severity from mild to severe, occur when a traumatic event, such as a fall, a traffic accident, or a blow, causes the brain to move rapidly within the skull, resulting in damage. Long-term consequences of TBI can include motor and cognitive deficits and emotional disturbances that result in a reduced quality of life and work productivity. Recovery from TBI can be challenging due to a lack of effective treatment options for repairing TBI-induced neural damage and alleviating functional impairments. Central nervous system (CNS) injury and disease are known to induce the activation of the small GTPase RhoA and its downstream effector Rho kinase (ROCK). Activation of this signaling pathway promotes cell death and the retraction and loss of neural processes and synapses, which mediate information flow and storage in the brain. Thus, inhibiting RhoA-ROCK signaling has emerged as a promising approach for treating CNS disorders. In this review, we discuss targeting the RhoA-ROCK pathway as a therapeutic strategy for treating TBI and summarize the recent advances in the development of RhoA-ROCK inhibitors.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Molecular Targeted Therapy , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Disease Models, Animal , Humans
7.
Elife ; 82019 08 28.
Article in English | MEDLINE | ID: mdl-31461398

ABSTRACT

Dendritic arbor architecture profoundly impacts neuronal connectivity and function, and aberrant dendritic morphology characterizes neuropsychiatric disorders. Here, we identify the adhesion-GPCR BAI1 as an important regulator of dendritic arborization. BAI1 loss from mouse or rat hippocampal neurons causes dendritic hypertrophy, whereas BAI1 overexpression precipitates dendrite retraction. These defects specifically manifest as dendrites transition from growth to stability. BAI1-mediated growth arrest is independent of its Rac1-dependent synaptogenic function. Instead, BAI1 couples to the small GTPase RhoA, driving late RhoA activation in dendrites coincident with growth arrest. BAI1 loss lowers RhoA activation and uncouples it from dendrite dynamics, causing overgrowth. None of BAI1's known downstream effectors mediates BAI1-dependent growth arrest. Rather, BAI1 associates with the Rho-GTPase regulatory protein Bcr late in development and stimulates its cryptic RhoA-GEF activity, which functions together with its Rac1-GAP activity to terminate arborization. Our results reveal a late-acting signaling pathway mediating a key transition in dendrite development.


Subject(s)
Angiogenic Proteins/metabolism , Cell Proliferation , Dendrites/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Mice , Rats
8.
Neuro Oncol ; 20(5): 655-665, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29112734

ABSTRACT

Background: Memantine has shown clinical utility in preventing radiation-induced cognitive impairment, but the mechanisms underlying its protective effects remain unknown. We hypothesized that abnormal glutamate signaling causes radiation-induced abnormalities in neuronal structure and that memantine prevents synaptic toxicity. Methods: Hippocampal cultures expressing enhanced green fluorescent protein were irradiated or sham-treated and their dendritic spine morphology assessed at acute (minutes) and later (days) times using high-resolution confocal microscopy. Excitatory synapses, defined by co-localization of the pre- and postsynaptic markers vesicular glutamate transporter 1 and postsynaptic density protein 95, were also analyzed. Neurons were pretreated with vehicle, the N-methyl-d-aspartate-type glutamate receptor antagonist memantine, or the glutamate scavenger glutamate pyruvate transaminase to assess glutamate signaling. For animal studies, Thy-1-YFP mice were treated with whole-brain radiotherapy or sham with or without memantine. Results: Unlike previously reported long-term losses of dendritic spines, we found that the acute response to radiation is an initial increase in spines and excitatory synapses followed by a decrease in spine/synapse density with altered spine dynamics. Memantine pre-administration prevented this radiation-induced synaptic remodeling. Conclusion: These results demonstrate that radiation causes rapid, dynamic changes in synaptic structural plasticity, implicate abnormal glutamate signaling in cognitive dysfunction following brain irradiation, and describe a protective mechanism of memantine.


Subject(s)
Abnormalities, Radiation-Induced/prevention & control , Dendritic Spines/drug effects , Gamma Rays/adverse effects , Hippocampus/drug effects , Memantine/pharmacology , Synapses/drug effects , Abnormalities, Radiation-Induced/etiology , Abnormalities, Radiation-Induced/pathology , Animals , Cells, Cultured , Dendritic Spines/pathology , Dendritic Spines/radiation effects , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/pathology , Hippocampus/radiation effects , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Synapses/radiation effects
9.
Sci Rep ; 7(1): 10689, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28878396

ABSTRACT

Traumatic brain injury (TBI) causes extensive neural damage, often resulting in long-term cognitive impairments. Unfortunately, effective treatments for TBI remain elusive. The RhoA-ROCK signaling pathway is a potential therapeutic target since it is activated by TBI and can promote the retraction of dendritic spines/synapses, which are critical for information processing and memory storage. To test this hypothesis, RhoA-ROCK signaling was blocked by RhoA deletion from postnatal neurons or treatment with the ROCK inhibitor fasudil. We found that TBI impairs both motor and cognitive performance and inhibiting RhoA-ROCK signaling alleviates these deficits. Moreover, RhoA-ROCK inhibition prevents TBI-induced spine remodeling and mature spine loss. These data argue that TBI elicits pathological spine remodeling that contributes to behavioral deficits by altering synaptic connections, and RhoA-ROCK inhibition enhances functional recovery by blocking this detrimental effect. As fasudil has been safely used in humans, our results suggest that it could be repurposed to treat TBI.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Cognition Disorders/etiology , Cognition Disorders/psychology , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors , Animals , Biomarkers , Brain Injuries, Traumatic/pathology , Dendrites/metabolism , Dendrites/pathology , Gene Deletion , Genotype , Immunohistochemistry , Male , Mice , Mice, Knockout , Models, Biological , Motor Activity , Neurons/metabolism , Prosencephalon/metabolism , Prosencephalon/pathology , Signal Transduction/drug effects , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
10.
Neurosci Lett ; 601: 4-10, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26003445

ABSTRACT

Synapses mediate information flow between neurons and undergo plastic changes in response to experience, which is critical for learning and memory. Conversely, synaptic defects impair information processing and underlie many brain pathologies. Rho-family GTPases control synaptogenesis by transducing signals from extracellular stimuli to the cytoskeleton and nucleus. The Rho-GTPases Rac1 and Cdc42 promote synapse development and the growth of axons and dendrites, while RhoA antagonizes these processes. Despite its importance, many aspects of Rho-GTPase signaling remain relatively unknown. Rho-GTPases are activated by guanine nucleotide exchange factors (GEFs) and inhibited by GTPase-activating proteins (GAPs). Though the number of both GEFs and GAPs greatly exceeds that of Rho-GTPases, loss of even a single GEF or GAP often has profound effects on cognition and behavior. Here, we explore how the actions of specific GEFs and GAPs give rise to the precise spatiotemporal activation patterns of Rho-GTPases in neurons. We consider the effects of coupling GEFs and GAPs targeting the same Rho-GTPase and the modular pathways that connect specific cellular stimuli with a given Rho-GTPase via different GEFs. We discuss how the creation of sharp borders between Rho-GTPase activation zones is achieved by pairing a GEF for one Rho-GTPase with a GAP for another and the extensive crosstalk between different Rho-GTPases. Given the importance of synapses for cognition and the fundamental roles that Rho-GTPases play in regulating them, a detailed understanding of Rho-GTPase signaling is essential to the progress of neuroscience.


Subject(s)
GTPase-Activating Proteins/metabolism , Synapses/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Brain Diseases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Neurons/metabolism , Signal Transduction
11.
Dev Biol ; 394(1): 39-53, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25128586

ABSTRACT

The small GTPases RhoA and Rac1 are key cytoskeletal regulators that function in a mutually antagonistic manner to control the migration and morphogenesis of a broad range of cell types. However, their role in shaping the cerebellum, a unique brain structure composed of an elaborate set of folia separated by fissures of different lengths, remains largely unexplored. Here we show that dysregulation of both RhoA and Rac1 signaling results in abnormal cerebellar ontogenesis. Ablation of RhoA from neuroprogenitor cells drastically alters the timing and placement of fissure formation, the migration and positioning of granule and Purkinje cells, the alignment of Bergmann glia, and the integrity of the basement membrane, primarily in the anterior lobules. Furthermore, in the absence of RhoA, granule cell precursors located at the base of fissures fail to undergo cell shape changes required for fissure initiation. Many of these abnormalities can be recapitulated by deleting RhoA specifically from granule cell precursors but not postnatal glia, indicating that RhoA functions in granule cell precursors to control cerebellar morphogenesis. Notably, mice with elevated Rac1 activity due to loss of the Rac1 inhibitors Bcr and Abr show similar anterior cerebellar deficits, including ectopic neurons and defects in fissure formation, Bergmann glia organization and basement membrane integrity. Together, our results suggest that RhoA and Rac1 play indispensable roles in patterning cerebellar morphology.


Subject(s)
Cerebellum/embryology , Morphogenesis/genetics , Neuropeptides/biosynthesis , rac1 GTP-Binding Protein/biosynthesis , rho GTP-Binding Proteins/genetics , Animals , Basement Membrane/physiology , Body Patterning/genetics , Cell Movement , Estrogen Antagonists/pharmacology , GTPase-Activating Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/physiology , Proto-Oncogene Proteins c-bcr/genetics , Signal Transduction , Tamoxifen/pharmacology , rhoA GTP-Binding Protein
12.
Dev Cell ; 29(6): 701-15, 2014 Jun 23.
Article in English | MEDLINE | ID: mdl-24960694

ABSTRACT

The small GTPase Rac1 orchestrates actin-dependent remodeling essential for numerous cellular processes including synapse development. While precise spatiotemporal regulation of Rac1 is necessary for its function, little is known about the mechanisms that enable Rac1 activators (GEFs) and inhibitors (GAPs) to act in concert to regulate Rac1 signaling. Here, we identify a regulatory complex composed of a Rac-GEF (Tiam1) and a Rac-GAP (Bcr) that cooperate to control excitatory synapse development. Disruption of Bcr function within this complex increases Rac1 activity and dendritic spine remodeling, resulting in excessive synaptic growth that is rescued by Tiam1 inhibition. Notably, EphB receptors utilize the Tiam1-Bcr complex to control synaptogenesis. Following EphB activation, Tiam1 induces Rac1-dependent spine formation, whereas Bcr prevents Rac1-mediated receptor internalization, promoting spine growth over retraction. The finding that a Rac-specific GEF/GAP complex is required to maintain optimal levels of Rac1 signaling provides an important insight into the regulation of small GTPases.


Subject(s)
Dendritic Spines/physiology , GTPase-Activating Proteins/physiology , Guanine Nucleotide Exchange Factors/metabolism , Proto-Oncogene Proteins c-bcr/physiology , Receptors, Eph Family/metabolism , Synapses/physiology , rac1 GTP-Binding Protein/metabolism , Animals , Blotting, Western , Electrophysiology , Endocytosis , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Immunoenzyme Techniques , Immunoprecipitation , Mice , Mice, Knockout , Neurites/metabolism , RNA, Small Interfering/genetics , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1
13.
PLoS One ; 8(6): e67015, 2013.
Article in English | MEDLINE | ID: mdl-23825607

ABSTRACT

The assembly of neuronal circuits during development requires the precise navigation of axons, which is controlled by attractive and repulsive guidance cues. In the developing spinal cord, ephrinB3 functions as a short-range repulsive cue that prevents EphA4 receptor-expressing corticospinal tract and spinal interneuron axons from crossing the midline, ensuring proper formation of locomotor circuits. Here we report that the small GTPase RhoA, a key regulator of cytoskeletal dynamics, is also required for ephrinB3/EphA4-dependent locomotor circuit formation. Deletion of RhoA from neural progenitor cells results in mice that exhibit a rabbit-like hopping gait, which phenocopies mice lacking ephrinB3 or EphA4. Consistent with this locomotor defect, we found that corticospinal tract axons and spinal interneuron projections from RhoA-deficient mice aberrantly cross the spinal cord midline. Furthermore, we determined that loss of RhoA blocks ephrinB3-induced growth cone collapse of cortical axons and disrupts ephrinB3 expression at the spinal cord midline. Collectively, our results demonstrate that RhoA is essential for the ephrinB3/EphA4-dependent assembly of cortical and spinal motor circuits that control normal locomotor behavior.


Subject(s)
Locomotion , Nerve Net/enzymology , Nerve Net/physiology , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Brain/cytology , Ephrin-B3/metabolism , Gene Knockout Techniques , Growth Cones/metabolism , Mice , Molecular Sequence Data , Nerve Net/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptor, EphA4/metabolism , Spinal Cord/cytology , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics
14.
J Biol Chem ; 287(9): 6830-9, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22223637

ABSTRACT

Using yeast-two hybrid screening followed by co-immunoprecipitation assay, we have found that the Lafora disease ubiquitin ligase malin interacts with dishevelled2, a key mediator of Wnt signaling pathway. Overexpression of malin enhances the degradation of dishevelled2 and inhibits Wnt signaling, which is evident from the down-regulation of ß-catenin target genes and the decrease in ß-catenin-mediated transcriptional activity. Partial knockdown of malin significantly increases the level of dishevelled2 and up-regulates Wnt signaling. Several malin mutants are found to be ineffective in degrading dishevelled2 and regulating the Wnt pathway. We have also found that malin enhances K48- and K63-linked ubiquitination of dishevelled2 that could lead to its degradation through both proteasome and autophagy. Altogether, our results indicate that malin regulates Wnt signaling pathway through the degradation of dishevelled2 and suggest possible deregulation of Wnt signaling in Lafora disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Phosphoproteins/metabolism , Wnt Signaling Pathway/physiology , Autophagy/physiology , Dishevelled Proteins , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Lafora Disease/pathology , Proteasome Endopeptidase Complex/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases , Ubiquitination/physiology , Up-Regulation/physiology , beta Catenin/metabolism
15.
Neurobiol Dis ; 40(3): 586-92, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20696245

ABSTRACT

E6 associated protein is an E3 ubiquitin ligase encoded by the gene Ube3a. Deletion or loss of function of the maternally inherited allele of Ube3a leads to Angelman syndrome. In the present study, we show that maternal loss of Ube3a (Ube3a(m-/p+)) in the mouse model leads to motor deficits that could be attributed to the dysfunction of the nigrostriatal pathway. The number of tyrosine hydroxylase positive neurons in the substantia nigra was significantly reduced in Ube3a(m-/p+) mice as compared to the wild type counterparts. The Ube3a(m-/p+) mice performed poorly in behavioural paradigms sensitive to nigrostriatal dysfunction. Even though the tyrosine hydroxylase staining was apparently the same in the striatum of both genotypes, the presynaptic and postsynaptic proteins were significantly reduced in Ube3a(m-/p+) mice. These findings suggest that the abnormality in the nigrostriatal pathway along with the cerebellum produces the observed motor dysfunctions in Ube3a(m-/p+) mice.


Subject(s)
Angelman Syndrome/physiopathology , Brain/physiopathology , Dopamine/metabolism , Neurons/pathology , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Behavior, Animal , Brain/metabolism , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Neurons/metabolism , Ubiquitin-Protein Ligases/genetics
16.
J Neurochem ; 110(6): 1955-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19645749

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder caused mainly because of the loss of dopaminergic neurons in the substantia nigra. Protein inclusions called Lewy bodies are the most common pathological hallmark of PD and other synucleinopathies. Because the main component of these inclusions is alpha-synuclein, aggregation of this protein is thought to be a key pathogenic event in this disease. In the present investigation we report that E6 associated protein (E6-AP), a HECT (homologous to E6-AP C-terminus) domain ubiquitin ligase is a component of Lewy bodies in post-mortem PD brain. In the cell culture model, we demonstrate that endogenous E6-AP colocalizes with alpha-synuclein in juxtanuclear aggregates. E6-AP is also recruited to the centrosome upon inhibition of the proteasome function suggesting its involvement in the degradation of misfolded proteins. Over-expression of E6-AP enhances the degradation of wild type as well as the mutant forms of alpha-synuclein in a proteasome-dependent manner. E6-AP also promotes the degradation of the more toxic oligomeric forms of alpha-synuclein. Our data suggests that E6-AP is involved in the clearance of alpha-synuclein.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/drug effects , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Brain/pathology , COS Cells , Chlorocebus aethiops , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lewy Bodies/metabolism , Mice , Mutation/genetics , Neuroblastoma , Parkinson Disease/pathology , Transfection/methods , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/pharmacology , alpha-Synuclein/genetics
17.
Biochem Biophys Res Commun ; 342(1): 184-90, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16472774

ABSTRACT

Huntington's disease (HD) is a familial neurodegenerative disorder caused by an abnormal expansion of CAG repeats in the coding region of huntingtin gene. A major hallmark of HD is the proteolytic production of N-terminal fragments of huntingtin containing polyglutamine repeats that form ubiquitinated aggregates in the nucleus and cytoplasm of the affected neurons. However, the mechanism by which the mutant huntingtin causes neurodegeneration is not well understood. Here, we found that oxidative stimuli enhance the polyglutamine-expanded truncated N-terminal huntingtin (mutant huntingtin) aggregation and mutant huntingtin-induced cell death. Oxidative stimuli also lead to rapid proteasomal dysfunction in the mutant huntingtin expressing cells as compared to normal glutamine repeat expressing cells. Overexpression of Cu/Zn superoxide dismutase (SOD1), Hsp40 or Hsp70 reverses the oxidative stress-induced proteasomal malfunction, mutant huntingtin aggregation, and death of the mutant huntingtin expressing cells. Finally, we show the higher levels of expression of SOD1 and DJ-1 in the mutant huntingtin expressing cells. Our result suggests that oxidative stress-induced proteasomal malfunction might be linked with mutant huntingtin-induced cell death.


Subject(s)
Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , Cell Death/drug effects , Cell Line , Gene Expression , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Huntington Disease/pathology , Mutation/genetics , Oncogene Proteins/metabolism , Oxidation-Reduction , Peptides/metabolism , Proteasome Inhibitors , Protein Binding , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Time Factors , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...